HSPA1A Antibody

Shipped with Ice Packs
In Stock

Description

Introduction

HSPA1A (Heat Shock Protein Family A Member 1A), also known as HSP70, is a stress-inducible molecular chaperone critical for maintaining protein homeostasis during cellular stress. Its antibody, a polyclonal or monoclonal immunoglobulin, is a widely used research tool to detect and quantify HSPA1A expression in various biological contexts, including cancer, immune modulation, and stress responses . This article synthesizes data from diverse sources to provide a comprehensive overview of the HSPA1A antibody, its mechanisms, applications, and clinical relevance.

Structure and Function of HSPA1A

HSPA1A is a 70-kDa protein encoded by the HSPA1A gene, located in the MHC class III region. It functions as a cytosolic chaperone, preventing protein aggregation and facilitating protein folding during stress. Beyond its cytosolic role, HSPA1A localizes to the plasma membrane of stressed or cancerous cells, where it interacts with lipids like phosphatidylserine (PS), enabling its membrane anchorage . This dual localization underscores its adaptive role in cellular stress and immune regulation.

Applications of the HSPA1A Antibody in Research

The antibody is employed in multiple experimental techniques:

ApplicationTechniqueKey Findings
Cancer BiologyWestern blot, IHCDetects HSPA1A overexpression in tumor cells and correlates with metastasis .
Autoimmune DiseasesELISA, Western blotSerum HSPA1A levels predict rheumatoid arthritis (RA) severity (AUC = 0.71) .
Immune ModulationFlow cytometry, IHCBinds Toll-like receptors (TLRs) on neutrophils, enhancing immune responses .

Methods of Detection and Antibody Specificity

Several antibodies targeting HSPA1A are available, differing in specificity and application:

AntibodyHostApplicationsReactivity
MAB1663MouseWestern blot, IHCHuman/mouse/rat HSPA1A
AF1663RabbitWestern blot, Simple WesternHuman/mouse/rat HSPA1A
PACO02105RabbitWestern blot, ELISAHuman HSPA1A (K246 site)

These antibodies exhibit minimal cross-reactivity with constitutive HSC70, ensuring specificity .

Clinical Relevance and Therapeutic Potential

HSPA1A is implicated in:

DiseaseRoleAntibody-Based Insights
CancerTumor growth, metastasisMembrane-bound HSPA1A activates immune cells and promotes survival .
RADisease severity markerElevated serum HSPA1A (>0.42 ng/mL) distinguishes RA from healthy controls .
NeurodegenerationProtein misfoldingHSPA1A sequesters misfolded proteins, mitigating oxidative stress .

A novel anti-HSPA1A huIgG1 antibody (ASY-77A) targets ADP-bound HSPA1A:peptide complexes, showing promise in preclinical cancer models .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days after receiving them. Delivery times may vary depending on the method of purchase and location. Please consult your local distributor for specific delivery timeframes.
Synonyms
HSPA1A antibody; HSP72 antibody; HSPA1 antibody; HSX70Heat shock 70 kDa protein 1A antibody; Heat shock 70 kDa protein 1 antibody; HSP70-1 antibody; HSP70.1 antibody
Target Names
HSPA1A
Uniprot No.

Target Background

Function
HSPA1A, also known as Heat Shock Protein 70 (HSP70), is a molecular chaperone that plays a crucial role in maintaining cellular proteostasis. It participates in a wide range of cellular processes, including:

• **Protein Folding and Transport:** HSPA1A assists in the proper folding of newly synthesized polypeptides and their subsequent transport to their designated locations within the cell.
• **Stress Response:** HSPA1A protects the cellular proteome from stress, preventing misfolding and aggregation of proteins, which can be detrimental to cellular function.
• **Misfolded Protein Degradation:** HSPA1A activates the proteolysis of misfolded proteins, ensuring their removal from the cell.
• **Protein Complex Formation and Dissociation:** HSPA1A facilitates the formation and dissociation of protein complexes, contributing to the dynamic nature of protein interactions within the cell.

HSPA1A is a key component of the protein quality control system, ensuring that proteins are correctly folded, refolded when necessary, and targeted for degradation when misfolded. This intricate regulation is achieved through a cycle involving ATP binding, ATP hydrolysis, and ADP release, which are modulated by co-chaperones.

Co-chaperones act as regulators of HSPA1A activity, influencing different stages of the ATPase cycle and exhibiting specific affinities for various substrates. Some co-chaperones promote the folding of certain substrates, while others trigger their degradation.

HSPA1A's affinity for polypeptides is directly influenced by its nucleotide-bound state. In the ATP-bound state, it exhibits a low affinity for substrate proteins. However, upon hydrolysis of ATP to ADP, HSPA1A undergoes a conformational change, increasing its affinity for substrate proteins. This continuous cycle of ATP hydrolysis and nucleotide exchange allows HSPA1A to bind and release substrates repeatedly.

Three primary types of co-chaperones interact with HSPA1A:

• **J-domain co-chaperones (HSP40s):** These co-chaperones stimulate ATPase hydrolysis by HSPA1A.
• **Nucleotide exchange factors (NEF) such as BAG1/2/3:** These co-chaperones facilitate the conversion of HSPA1A from the ADP-bound to the ATP-bound state, promoting substrate release.
• **TPR domain chaperones such as HOPX and STUB1:** These co-chaperones play a crucial role in various cellular processes.

During cellular stress, HSPA1A maintains protein homeostasis by promoting both protein refolding and degradation. Its acetylation/deacetylation state determines its functional role, either in protein refolding or degradation, by regulating the binding of co-chaperones HOPX and STUB1. During the initial stress response, the acetylated form of HSPA1A binds to HOPX, assisting in chaperone-mediated protein refolding. Subsequently, HSPA1A is deacetylated and binds to the ubiquitin ligase STUB1, promoting ubiquitin-mediated protein degradation.

HSPA1A plays a vital role in various cellular processes, including:

• **Centrosome Integrity:** HSPA1A regulates centrosome integrity during mitosis, ensuring the formation of a functional mitotic centrosome that supports the assembly of a bipolar mitotic spindle.
• **TGF-beta Signaling:** HSPA1A enhances STUB1-mediated SMAD3 ubiquitination and degradation, leading to inhibition of TGF-beta signaling.
• **Immune Response:** HSPA1A is essential for STUB1-mediated ubiquitination and degradation of FOXP3 in regulatory T-cells (Treg) during inflammation.
• **Heat Shock Response:** HSPA1A negatively regulates heat shock-induced HSF1 transcriptional activity during the attenuation and recovery phase of the heat shock response.
• **Protein Degradation:** HSPA1A is involved in the clearance of misfolded PRDM1/Blimp-1 proteins, sequestering them in the cytoplasm and promoting their association with SYNV1/HRD1, leading to proteasomal degradation.

**Microbial Infection:** In cases of rotavirus A infection, HSPA1A acts as a post-attachment receptor for the virus, facilitating its entry into the cell.
Gene References Into Functions
  1. The binding of IL-5 to IL-5Ralpha receptors enhances angiogenic responses by stimulating the expression of HSPA1A via the eNOS signaling pathway. PMID: 28317868
  2. Downregulation of HSPA1A impairs mesenchymal stem cell osteogenic and chondrogenic differentiation. PMID: 29323151
  3. HSPA1A overexpression promotes lipid accumulation in hepatocytes. PMID: 29631603
  4. Studies demonstrate that HSP72 inhibits HDACi-induced apoptosis in the Jurkat cell line. PMID: 29395577
  5. In conclusion, HSP70 modulates NF-kappaB activation in alveolar macrophages of TB patients, through inhibiting IkappaB-alpha phosphorylation or acting as a chaperon molecule to prevent NF-kappaB binding to the target genes by facilitating degradation. The upregulated HSP70 may suppress the release of pro-inflammatory cytokines during active pulmonary tuberculosis infection, and prevent overwhelming tissue damage. PMID: 28450725
  6. HSPA6 and HSPA1A contribute to the protection of differentiated human neuronal cells from cellular stress. PMID: 29090408
  7. Ultramarathon running caused a substantial increase in eHsp72 concentration, but probiotic + glutamine supplementation did not affect eHsp72 levels. PMID: 28460195
  8. uHSP72 may be considered as a novel potential diagnostic biomarker for the early detection of Diabetic nephropathy (DN). Moreover, these data support the pivotal role of NLRP3 in the development and progression of DN. PMID: 28631886
  9. The G allele of rs1008438G>T of HSPA1A may be a protective factor for cervical cancer among ethnic Han Chinese from Yunnan. PMID: 29188629
  10. Measurable HSP72 was not associated with graft versus host disease following allogeneic hematopoietic cell transplantation. PMID: 27020764
  11. Studies demonstrated that ovarian cancer cells isolated from patients with type II tumors released high levels of immunosuppressive cytokines (i.e., interleukin 10 and transforming growth factor beta) and HspA1A in vitro. PMID: 26868087
  12. This study suggests that logotherapy affects the expression of cortisol, BDI, and pain scales in advanced cervical cancer patients, and that it does not affect the expression of HSP70. PMID: 27644267
  13. Data suggest that two putative NEF (nucleotide exchange factors) orthologs, GRPEL1 and GRPEL2, modulate function of mitochondrial HSP70 (mtHSP70); both GRPEL1 and GRPEL2 associate with mtHSP70 as hetero-oligomeric subcomplex and regulate mtHSP70 transport. (GRPEL = mitochondrial GrpE-like protein; HSP70 = heat-shock protein 70). PMID: 28848044
  14. High HSP72 expression is associated with Cluster Amplified Centrosomes in cancer. PMID: 28720575
  15. mRNA levels of HSP family members (HSP70B', HSP72, HSP40/DNAJ, and HSP20/CRYAB) are upregulated by the intracellular MMP3 overload. PMID: 27206651
  16. Data suggest that both ATP- and peptide-binding domains of HSPA1A can form complexes with an AU-rich element in VEGFA mRNA in vitro; only peptide-binding domain can recover cellular VEGFA mRNA in ribonucleoprotein immunoprecipitation; RNA-binding and mRNA-stabilizing functions of HSPA1A are independent of its protein chaperone cycle. (HSPA1A = heat shock 70 kDa protein 1A; VEGFA = vascular endothelial growth factor A). PMID: 28679534
  17. It has been demonstrated that HSPB8-BAG3-HSP70 ensures the functionality of stress granules and restores proteostasis by targeting defective ribosomal products for degradation. PMID: 27570075
  18. The rs2763979 locus of the HSP70 genes may be associated with susceptibility to noise-induced hearing loss (NIHL) in Chinese individuals, and other HSP70 genes may also be susceptibility genes for NIHL, but the results must be further replicated in additional independent sample sets. PMID: 28182740
  19. These results suggest that NFkappaB engaged with the kappaB motif on the promoter cooperates in Hsp70A1A activation under heat shock in human cells as part of a DNA-break repair complex including DNA-PK and PARP-1. PMID: 28099440
  20. Systematic proteomic identification of the heat shock proteins that interact with estrogen receptor alpha and biochemical characterization of the ERalpha-hsp70 interaction has been reported. PMID: 27483141
  21. Epidermal Hsp70-1A contributes to the diversity of skin color by regulating the amount of melanin synthesized in melanocytes and modulating autophagic melanosome degradation in keratinocytes. PMID: 27094592
  22. Extracellular Hsp72 immediately post-exercise decreased back to baseline levels by 1 h post-exercise, but cellular Hsp72 expression continued to rise and remained elevated 24 h post-exercise. These data suggest that in addition to the classic physiological biomarkers of exercise heat stress, using cellular Hsp72 as an indicator of lasting effects of stress into recovery may be most appropriate for determining long-term ef PMID: 26643874
  23. HSPA1A and HSPA8 have roles in parturition through stimulating immune inflammatory and estrogen response. PMID: 28025138
  24. Data show that BAG2 Inhibits CHIP-Mediated HSP72 ubiquitination in aged cells. PMID: 28042827
  25. Indicates increased expression levels of heat shock proteins 90 and 70 and glucose related protein 78 levels in medullary thyroid carcinoma. PMID: 28038712
  26. The cardioprotective effect of 40-60 g/d of alcohol consumption could be due in part, to increased intracellular HSPA1A, a potent anti-inflammatory protein. Excessive intake of alcohol increases antibodies anti-Hsp60, stimulating proinflammatory cytokines. This fact may explain the mortality from cardiovascular disease in heavy drinkers. PMID: 26902796
  27. There is a direct correlation between plasma HSPA1A and PAI-1 levels in patients with diabetes, which is lost when they develop albuminuria. PMID: 26637413
  28. The present study revealed that salivary extracellular HSP70 significantly increased at 4 h after the 59 min of intense exercise in sedentary male subjects and correlated with resting salivary secretory immunoglobulin A (SIgA) levels at rest. PMID: 26608509
  29. HSP70-2 (+1267A/G) gene polymorphism was associated with Henoch-Schonlein purpura in children. PMID: 26547206
  30. HSP72 blocks fibroblast activation and proliferation in renal fibrosis via targeting the STAT3 pathway and may serve as a novel therapeutic agent for chronic kidney disease regardless of the etiology. PMID: 26851345
  31. Studies suggest that heat shock protein 72/70 (Hsp70 proteins) are beneficial to the patient in slowing the onset of neurodegenerative disorders. PMID: 26450908
  32. P53 could be used to distinguish early HCC from advanced hepatocellular carcinoma, but HSP70 cannot. PMID: 26494212
  33. Cytokines, not being influenced by HSP72 polymorphisms, cortisol, or illness severity. Gln may depress l/mHSP72 after LPS exposure and enhance them after HS induction, but it may not affect early induced HSP72 mRNA. PMID: 26550577
  34. Heat acclimation reduces physiological strain, and the transcription of HSP72, but not HSP90alpha mRNA in acute normobaric hypoxia. PMID: 26205540
  35. Hsp72 (HSPA1A) prevents h-IAPP aggregation and toxicity. PMID: 26960140
  36. HSPA1A (rs1043618) is associated with a decreased risk of idiopathic pulmonary fibrosis in a Mexican population. PMID: 26496868
  37. Lysine methylation of HSPA1 differs between metastatic breast and ovarian carcinoma. PMID: 26448330
  38. Together, these results implicate HSP70-1A as a novel angiogenic regulator. PMID: 26657847
  39. The aim of this study was to demonstrate the effects of 6-week low-intensity training on changes in indicators of aerobic capacity and on HSPA1A, HSPB1, and LDHb expression in white blood cells in high level rowers. PMID: 26214432
  40. BDNF, APOE, and HSP70-1 genes, but not GRIN2B, might be associated with a risk of POAG occurrence in the Polish population. PMID: 25893192
  41. Leukocyte Hsp72 mRNA was increased immediately after exercise following downhill running compared to flat running and in hot compared to temperate conditions. PMID: 25722377
  42. Equal Hsp72 mRNA increases occurring from consistent, reduced, or increased endogenous strain following short-term heat acclimation and long-term heat acclimation suggest that transcription occurs following attainment of sufficient endogenous criteria. PMID: 25943677
  43. Higher levels of plasma Hsp70 and lower levels of plasma Hsp27 might be associated with an increased risk of COPD among coal workers. PMID: 25620081
  44. Serum HSPA1A levels correlate with disease status in rheumatoid arthritis. PMID: 25739548
  45. This is indicative of improved tolerance and ability to cope with the hypoxic insult, potentially mediated in part by increased basal reserves of HSP72. PMID: 25874231
  46. Data indicate that the smallest average tumor weight was present in the AdSurp-heat shock 70kDa protein (Hsp70)+CIK treatment group. PMID: 25473902
  47. Results report high resolution crystal structure of the substrate-bound human HSP70-substrate-bound domain and particularly the alpha and beta loops. PMID: 25058147
  48. HSP72 preserves renal function in I/R injury through its antiapoptotic effects, which act by suppressing mitochondrial Smac/Diablo release and preserving XIAP protein content. PMID: 25394481
  49. The results demonstrate a key role for inducible HSP70 in aiding the processing and hindering the accumulation of misfolded PMP22, which in turn alleviates proteotoxicity within the cells. PMID: 25694550
  50. Nek6 facilitates association of Hsp72 with the mitotic spindle, where it promotes stable K-fiber assembly through recruitment of the ch-TOG-TACC3 complex. PMID: 25940345

Show More

Hide All

Database Links

HGNC: 5232

OMIM: 140550

KEGG: hsa:3303

STRING: 9606.ENSP00000364802

UniGene: Hs.274402

Protein Families
Heat shock protein 70 family
Subcellular Location
Cytoplasm. Nucleus. Cytoplasm, cytoskeleton, microtubule organizing center, centrosome. Secreted. Note=Localized in cytoplasmic mRNP granules containing untranslated mRNAs.

Customer Reviews

Overall Rating 5.0 Out Of 5
,
B.A
By Anonymous
★★★★★

Applications : Immunohistochemical staining

Sample type: Cells

Review: Specimens were washed twice with TPBS buffer and incubated with H2O2 for 5–10 minutes to block peroxidase enzyme activity. Specimens were then incubated with diluted rabbit primary antibody (CSB-PA556911,Cusabio Technology,China), followed by incubation with the HRP-labeled polymer.

Q&A

What is HSPA1A and what are its alternative nomenclatures in scientific literature?

HSPA1A (heat shock protein family A member 1A) is a 70.1 kilodalton molecular chaperone that regulates the survival of stressed and cancer cells. In scientific literature, HSPA1A may also be referred to as HSP70-1A, HSP70I, HEL-S-103, HSP70-1, HSP70-1A, HSP70-2, and heat shock 70 kDa protein 1A . This protein plays critical roles in cellular stress responses and has both cytosolic and membrane-associated functions. When designing experiments targeting HSPA1A, researchers should be aware of these alternative nomenclatures to ensure comprehensive literature searches and proper antibody selection. Mouse and rat orthologs may also be found based on gene name, which is important for cross-species research considerations .

What cell lines are commonly used for HSPA1A antibody validation and expression studies?

For HSPA1A antibody validation and expression studies, human cell lines such as HEK293 (Human embryonic kidney cells; ATCC® CRL-1573™) and HeLa (Henrietta Lacks' 'Immortal' cells; ATCC® CCL-2™) are frequently employed . When working with these cell lines, they should be maintained in a humidified 5% CO₂ atmosphere at 37°C. HEK293 cells are typically cultured in DMEM supplemented with 10% fetal bovine serum, 2 mM L-glutamine, and penicillin-streptomycin, while HeLa cells require MEM with the same supplements plus 0.1 mM non-essential amino acids and sodium pyruvate . For immunomodulatory studies, differentiated U937 macrophages (ATCC CRL-1593.2) maintained in RPMI 1640 with 10% FBS are also utilized to investigate HSPA1A interactions with various toll-like receptors .

What are the standard applications for which HSPA1A antibodies are validated?

HSPA1A antibodies are validated for numerous research applications, with Western Blot (WB) being the most universally supported application across different antibody products . Other common applications include Enzyme-Linked Immunosorbent Assay (ELISA), Immunofluorescence (IF), Immunohistochemistry (IHC), Immunoprecipitation (IP), and Flow Cytometry (FCM) . When selecting an HSPA1A antibody for a specific application, researchers should verify that the antibody has been validated for that particular technique. For instance, some antibodies like the HSPA1A Polyclonal Antibody offered by multiple suppliers are validated for WB and ELISA, while others such as specific monoclonal antibodies from Creative Biolabs are validated for additional techniques including IP and FCM . The choice between polyclonal and monoclonal antibodies should be based on the specific experimental requirements and the need for specificity versus epitope recognition.

How should researchers optimize Western blot protocols for HSPA1A detection?

For optimal Western blot detection of HSPA1A, researchers should consider using specific validated antibodies such as the HSPA1A (HSP70) clone #C92F3A-5 from Enzo Life Sciences, which detects a protein band of approximately 74 kDa for both native and overexpressed HSPA1A . An effective protocol includes overnight (~16 h) incubation with primary antibodies at 4°C with constant rotation at a dilution of 1:1000 . Appropriate controls are essential: β-actin (approximately 42 kDa) serves as a cytosolic loading control, while Na+/K+ ATPase α (ATP1A1) (approximately 112 kDa) functions as a plasma membrane loading control . This dual control strategy is particularly important when studying HSPA1A's differential localization between cytosolic and membrane compartments. Researchers should also optimize sample preparation based on their specific cell types, considering that HSPA1A's expression and localization may vary under different stress conditions or in different cancer cell lines.

How can researchers effectively distinguish between cytosolic and membrane-associated HSPA1A in experimental settings?

To effectively distinguish between cytosolic and membrane-associated HSPA1A, researchers should implement a systematic subcellular fractionation protocol followed by Western blot analysis using specific loading controls. The cytosolic fraction should be verified using β-actin antibody (detecting a 42 kDa band), while the plasma membrane fraction can be confirmed using Na+/K+ ATPase α (ATP1A1) antibody (detecting a 112 kDa band) . For quantitative analysis, it is recommended to perform densitometry on Western blots and normalize HSPA1A levels to their respective compartment-specific loading controls.

Since HSPA1A lacks known membrane localization signals yet binds to lipids like phosphatidylserine (PS) and monophosphorylated phosphoinositides (PIPs), researchers investigating membrane localization mechanisms should consider incorporating lipid binding assays . Immunofluorescence microscopy with membrane-specific stains or plasma membrane biotinylation assays provide complementary approaches to verify membrane localization. When studying cancer cells, it's important to note that HSPA1A's membrane presentation is often enhanced compared to normal cells, potentially serving as a biomarker for malignant transformation or stress response.

What are the critical considerations when investigating HSPA1A interactions with Toll-Like Receptors (TLRs)?

When investigating HSPA1A interactions with TLRs, researchers should implement a multi-faceted experimental approach that accounts for potential cross-talk between different receptor systems. Flow cytometry analysis represents a powerful technique for studying these interactions, where differentiated cells (such as U937 macrophages) at a concentration of 2.5×10⁵ cells/ml can be treated with or without 1000 ng/ml of HSPA1A for approximately 6 hours . Cells should be immunostained with receptor-specific antibodies such as Anti-TLR2 (0.1 mg/ml), Anti-TLR4 (0.1 mg/ml), Anti-TLR5 (0.1 mg/ml), or Anti-TLR7 (0.2 mg/ml) conjugated to PE (available from suppliers like BD BioSciences) .

To assess cell viability during these experiments, propidium iodide staining (10 μg of 50 μg/ml solution) for 1 hour at 4°C in dark conditions should be employed, and flow cytometric analysis should include only viable cells . For functional readouts of these interactions, ELISA assays measuring cytokine secretion (TNF-α, IL-1β, IL-10) provide valuable insights. Blocking peptide experiments, where cells are pre-incubated with TLR-specific blocking peptides before HSPA1A exposure, can help determine the specific contribution of each receptor to the observed immunomodulatory effects . Data analysis should be performed using appropriate statistical software such as GraphPad Prism, with results presented as mean ± SD from at least three independent experiments with three replicates each.

How do post-translational modifications affect HSPA1A antibody recognition and what techniques can address this challenge?

Post-translational modifications (PTMs) of HSPA1A, including phosphorylation, acetylation, and ubiquitination, can significantly impact antibody recognition and biological function. Research indicates that phosphorylation at Y41 is particularly significant, as evidenced by the availability of phospho-specific antibodies targeting this modification . To comprehensively investigate HSPA1A PTMs, researchers should employ a combination of approaches including phospho-specific antibodies, such as Phospho-HSPA1A/HSPA1B (Y41) Antibody, alongside the standard HSPA1A antibodies to compare total versus modified protein levels .

Mass spectrometry-based proteomics provides the most comprehensive approach for identifying novel or less characterized PTMs. For confirmation of specific modifications, researchers can use immunoprecipitation with general HSPA1A antibodies followed by Western blotting with modification-specific antibodies. When studying phosphorylation dynamics, treatment with phosphatase inhibitors during sample preparation is crucial. Additionally, incorporating PTM-mimicking or PTM-deficient mutants in cell models can provide functional insights into the role of specific modifications on HSPA1A localization, particularly its transition between cytosolic and membrane-associated states. Researchers should be aware that certain antibodies may have reduced affinity for heavily modified HSPA1A, potentially leading to false negative results in heavily stressed or pathological samples.

What methodological approaches should be used to investigate the role of HSPA1A in tumor therapy resistance?

To investigate HSPA1A's role in tumor therapy resistance, researchers should implement a comprehensive methodological workflow that examines both membrane-bound (mHSPA1A) and extracellular (eHSPA1A) forms of the protein, as these have been implicated in immunomodulatory functions and therapy resistance . Cell culture experiments should compare therapy-sensitive and therapy-resistant cancer cell lines, examining differential HSPA1A expression, localization, and secretion patterns using a combination of Western blotting, flow cytometry, and ELISA techniques.

For in vitro therapy resistance models, researchers can establish drug-resistant cancer cell lines through gradual exposure to increasing concentrations of relevant therapeutic agents, followed by comparative analysis of HSPA1A expression and localization between parental and resistant lines. Functional studies should include siRNA or CRISPR-Cas9 mediated knockdown/knockout of HSPA1A, coupled with assessment of therapy sensitivity using cell viability, apoptosis, and clonogenic assays. To specifically address the role of membrane-associated HSPA1A, cell-impermeable HSPA1A-targeting antibodies or peptides can be employed to block its extracellular functions without affecting intracellular activities.

Because HSPA1A interacts with PIPs and phosphatidylserine in the plasma membrane , manipulating these lipids using specific inhibitors or lipidomic approaches can provide insights into the mechanisms of HSPA1A membrane translocation in therapy-resistant cells. Additionally, co-culture experiments with immune cells can help decipher how mHSPA1A and eHSPA1A modulate anti-tumor immune responses, potentially contributing to immunotherapy resistance. Data from these experiments should be correlated with clinical outcomes in patient samples to establish translational relevance.

What controls are essential when validating a new HSPA1A antibody for research applications?

When validating a new HSPA1A antibody, implementing a comprehensive set of positive and negative controls is crucial for ensuring specificity and reliability. Essential positive controls include recombinant HSPA1A protein for Western blot and ELISA applications, and cell lines known to express high levels of HSPA1A, such as heat-shocked HEK293 or HeLa cells . Negative controls should include HSPA1A-knockout or HSPA1A-knockdown cell lines generated using CRISPR-Cas9 or siRNA technology, respectively.

For cross-reactivity assessment, researchers should test the antibody against other HSP70 family members, particularly HSPA1B which shares high sequence homology with HSPA1A. This is especially important when using polyclonal antibodies, which may recognize multiple epitopes. Additionally, peptide competition assays, where the antibody is pre-incubated with excess immunizing peptide before application, can confirm binding specificity. For immunocytochemistry or immunohistochemistry applications, secondary antibody-only controls are necessary to evaluate background staining.

When validating antibodies for detecting specific HSPA1A localizations, such as membrane-associated HSPA1A, appropriate subcellular fractionation controls should be employed, including Na+/K+ ATPase α (ATP1A1) for plasma membrane fractions and β-actin for cytosolic fractions . Documentation of these validation experiments should include images of complete Western blots showing all bands detected by the antibody, and quantitative assessments of specificity and sensitivity across different experimental conditions.

How can researchers address discrepancies in HSPA1A antibody results between different experimental techniques?

Discrepancies in HSPA1A antibody results between different experimental techniques often stem from technique-specific limitations and epitope accessibility issues. To address these challenges, researchers should first verify that the antibody is validated specifically for each technique being employed, as antibodies that perform well in Western blot may not be suitable for immunoprecipitation or flow cytometry . When discrepancies occur, employing multiple antibodies targeting different epitopes of HSPA1A can help confirm results and identify potential epitope-specific artifacts.

Sample preparation differences represent a major source of variation between techniques. For instance, harsh detergents used in Western blotting may efficiently extract HSPA1A from all cellular compartments, while gentler fixation methods for immunofluorescence may preserve only specific pools of the protein. To address this, researchers should systematically compare different sample preparation protocols while maintaining consistent antibody concentrations and incubation conditions.

Post-translational modifications can also contribute to discrepancies by altering epitope accessibility. In such cases, using modification-specific antibodies, such as Phospho-HSPA1A/HSPA1B (Y41) Antibody , alongside total HSPA1A antibodies can help identify whether modifications are responsible for the observed differences. Additionally, for quantitative comparisons between techniques, appropriate calibration standards and loading controls should be included, and results should be normalized using technique-specific methods. When reporting discrepancies in the literature, researchers should clearly document all methodological details and consider the biological implications of technique-dependent results rather than simply dismissing them as technical artifacts.

What experimental design considerations are important when studying HSPA1A interactions with immune cell receptors?

When studying HSPA1A interactions with immune cell receptors such as TLRs, careful experimental design is essential to distinguish direct protein-receptor interactions from downstream signaling events. For cell-based interaction studies, differentiated U937 macrophages provide a valuable model system, as they express multiple TLRs that have been implicated in HSPA1A recognition . Experiments should include appropriate positive controls such as known TLR ligands (e.g., LPS for TLR4) to confirm receptor functionality.

A comprehensive experimental design should incorporate both binding and functional studies. Direct binding can be assessed through co-immunoprecipitation of HSPA1A with target receptors, while surface plasmon resonance or microscale thermophoresis can provide quantitative binding kinetics. For functional studies, cytokine secretion assays measuring TNF-α, IL-1β, and IL-10 production following HSPA1A stimulation offer valuable insights into receptor activation .

Receptor specificity should be systematically evaluated using receptor-specific blocking peptides or antibodies against TLR2, TLR4, TLR5, and TLR7, applied prior to HSPA1A exposure . For each receptor-blocking condition, dose-response experiments should be conducted to determine optimal blocking concentrations and potential synergistic effects between receptors. Time-course studies ranging from 1-24 hours post-stimulation can help distinguish between early and late immune response signatures.

To control for potential endotoxin contamination in recombinant HSPA1A preparations, which could confound TLR4-dependent responses, researchers should include polymyxin B controls and use endotoxin-free protein preparation protocols. Additionally, experiments comparing wild-type HSPA1A with mutant variants lacking specific domains can help identify the structural determinants of receptor interactions. All results should be statistically analyzed using appropriate methods such as ANOVA with post-hoc tests, with data presented as mean ± SD from at least three independent experiments with three replicates each .

How can HSPA1A antibodies be employed in cancer research and what methodological considerations are important?

HSPA1A antibodies serve as powerful tools in cancer research due to the protein's differential expression, localization, and function in malignant versus normal cells. For translational cancer research, immunohistochemistry with HSPA1A antibodies can help evaluate expression patterns across tumor types and correlate with clinical outcomes. Researchers should employ tissue microarrays with multiple tumor samples alongside matched normal tissues, using antibodies validated specifically for IHC applications . Scoring systems should be standardized, ideally using digital image analysis to quantify staining intensity and subcellular localization patterns.

Flow cytometry with HSPA1A antibodies offers a method to identify and potentially isolate cancer cell subpopulations with membrane-localized HSPA1A, which has been implicated in therapy resistance . This approach requires non-permeabilized cells and surface-specific staining protocols to distinguish membrane-bound from intracellular HSPA1A. For therapeutic development, cell-impermeable HSPA1A-targeting antibodies may selectively target cancer cells with elevated surface HSPA1A expression while sparing normal cells with predominantly intracellular HSPA1A.

Co-immunoprecipitation studies using HSPA1A antibodies can identify cancer-specific interaction partners that may represent novel therapeutic targets. These experiments should include appropriate controls for antibody specificity and consider potential differences in HSPA1A interactome between cytosolic and membrane-associated protein pools. Additionally, HSPA1A antibodies conjugated to nanoparticles or imaging agents may serve as cancer-specific diagnostic tools, exploiting the enhanced membrane localization of HSPA1A in malignant cells . As with all cancer biomarker studies, validation across multiple patient cohorts is essential before clinical application of HSPA1A-based diagnostics or therapeutics.

What methodological approaches can be used to study the interaction between HSPA1A and membrane lipids?

Liposome binding assays using synthetic liposomes with defined lipid compositions can quantitatively assess HSPA1A binding preferences. These experiments should include liposomes containing varying concentrations of PS and PIPs, alongside control liposomes lacking these lipids. For cellular studies, pharmacological manipulation of PIP levels using specific kinase or phosphatase inhibitors, followed by assessment of HSPA1A membrane localization through fractionation and Western blotting, can establish causative relationships between specific lipids and HSPA1A membrane association.

Advanced microscopy techniques, including total internal reflection fluorescence (TIRF) microscopy with fluorescently labeled HSPA1A, can visualize real-time interactions with membrane lipids in living cells. For molecular-level insights, molecular dynamics simulations based on HSPA1A structure and lipid binding domains can predict interaction interfaces and guide experimental design. Additionally, mass spectrometry-based lipidomics of HSPA1A-containing membrane fractions can identify the full spectrum of lipids associated with HSPA1A in different cellular contexts.

Researchers should also consider the impact of lipid oxidation and membrane fluidity on HSPA1A binding, particularly in stress and disease conditions where membrane properties may be altered. Comparative studies between normal and cancer cells may reveal disease-specific alterations in HSPA1A-lipid interactions that could inform therapeutic strategies targeting this protein's membrane localization.

How can researchers develop and validate HSPA1A antibodies for therapeutic applications?

Developing HSPA1A antibodies for therapeutic applications requires a methodological pipeline that extends beyond traditional research antibody validation. Initial steps should focus on generating antibodies with high specificity for HSPA1A over other HSP70 family members, particularly HSPA1B, using either hybridoma technology for monoclonal antibodies or phage display for recombinant antibodies. These antibodies should be rigorously tested for cross-reactivity against a panel of HSP70 family proteins using techniques such as ELISA and Western blotting.

For cancer therapy applications targeting membrane-associated HSPA1A, antibodies should be screened for preferential binding to membrane-localized versus intracellular HSPA1A. This can be achieved through flow cytometry on non-permeabilized cells expressing varying levels of surface HSPA1A. Additionally, therapeutic antibodies should be evaluated for their ability to induce antibody-dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC) against cancer cells expressing surface HSPA1A.

Functional screening should assess whether candidate antibodies can block HSPA1A interactions with immune cell receptors like TLRs, potentially modulating immunosuppressive effects in the tumor microenvironment . Antibodies showing promise in these initial screens should undergo humanization to reduce immunogenicity, followed by affinity maturation to optimize target binding.

Preclinical validation should include pharmacokinetic and biodistribution studies in relevant animal models, with particular attention to tumor penetration and retention. Toxicology studies must evaluate potential off-target effects on normal tissues expressing HSPA1A. Finally, combination studies with standard chemotherapies or immunotherapies can assess whether HSPA1A-targeting antibodies enhance treatment efficacy by overcoming therapy resistance mechanisms linked to membrane-associated HSPA1A . Throughout this development pipeline, researchers should maintain detailed documentation of all validation steps to support potential clinical translation.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.